Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shaoling Huang is active.

Publication


Featured researches published by Shaoling Huang.


Advanced Drug Delivery Reviews | 2008

Liposomes in ultrasonic drug and gene delivery.

Shaoling Huang

Liposome-based drug and gene delivery systems have potential for significant roles in a variety of therapeutic applications. Recently, liposomes have been used to entrap gas and drugs for ultrasound-controlled drug release and ultrasound-enhanced drug delivery. Echogenic liposomes have been produced by different preparation methods, including lyophilization, pressurization, and biotin-avidin binding. Presently, significant in vivo applications of liposomal ultrasound-based drug and gene delivery are being made in cardiac disease, stroke and tumor therapy. Translation of these vehicles into the clinic will require a better understanding of improved physical properties to avoid rapid clearance, as well as of possible side effects, including those of the ultrasound. The aim of this review is to provide orientation for new researchers in the area of ultrasound-enhanced liposome drug and gene delivery.


Thrombosis Research | 2009

Ultrasound-enhanced thrombolysis with tPA-loaded echogenic liposomes

George J. Shaw; Jason M. Meunier; Shaoling Huang; Christopher J. Lindsell; David D. McPherson; Christy K. Holland

BACKGROUND AND PURPOSE Currently, the only FDA-approved therapy for acute ischemic stroke is the administration of recombinant tissue plasminogen activator (tPA). Echogenic liposomes (ELIP), phospholipid vesicles filled with gas and fluid, can be manufactured to incorporate tPA. Also, transcranial ultrasound-enhanced thrombolysis can increase the recanalization rate in stroke patients. However, there is little data on lytic efficacy of combining ultrasound, echogenic liposomes, and tPA treatment. In this study, we measure the effects of pulsed 120-kHz ultrasound on the lytic efficacy of tPA and tPA-incorporating ELIP (t-ELIP) in an in-vitro human clot model. It is hypothesized that t-ELIP exhibits similar lytic efficacy to that of rt-PA. METHODS Blood was drawn from 22 subjects after IRB approval. Clots were made in 20-microL pipettes, and placed in a water tank for microscopic visualization during ultrasound and drug treatment. Clots were exposed to combinations of [tPA]=3.15 microg/ml, [t-ELIP]=3.15 microg/ml, and 120-kHz ultrasound for 30 minutes at 37 degrees C in human plasma. At least 12 clots were used for each treatment. Clot lysis over time was imaged and clot diameter was measured over time, using previously developed imaging analysis algorithms. The fractional clot loss (FCL), which is the decrease in mean clot width at the end of lytic treatment, was used as a measure of lytic efficacy for the various treatment regimens. RESULTS The fractional clot loss FCL was 31% (95% CI: 26-37%) and 71% (56-86%) for clots exposed to tPA alone or tPA with 120 kHz ultrasound. Similarly, FCL was 48% (31-64%) and 89% (76-100%) for clots exposed to t-ELIP without or with ultrasound. CONCLUSIONS The lytic efficacy of tPA containing echogenic liposomes is comparable to that of tPA alone. The addition of 120 kHz ultrasound significantly enhanced lytic treatment efficacy for both tPA and t-ELIP. Liposomes loaded with tPA may be a useful adjunct in lytic treatment with tPA.


Journal of the American College of Cardiology | 2009

Nitric Oxide Loaded Echogenic Liposomes for Nitric Oxide Delivery and Inhibition of Intimal Hyperplasia

Shaoling Huang; Patrick Kee; Hyunggun Kim; Melanie R. Moody; Stephen M. Chrzanowski; Robert C. MacDonald; David D. McPherson

OBJECTIVES We sought to develop a new bioactive gas-delivery method by the use of echogenic liposomes (ELIP) as the gas carrier. BACKGROUND Nitric oxide (NO) is a bioactive gas with potent therapeutic effects. The bioavailability of NO by systemic delivery is low with potential systemic effects. METHODS Liposomes containing phospholipids and cholesterol were prepared by the use of a new method, freezing under pressure. The encapsulation and release profile of NO from NO-containing ELIP (NO-ELIP) or a mixture of NO/argon (NO/Ar-ELIP) was studied. The uptake of NO from NO-ELIP by cultured vascular smooth muscle cells (VSMCs) both in the absence and presence of hemoglobin was determined. The effect of NO-ELIP delivery to attenuate intimal hyperplasia in a balloon-injured artery was determined. RESULTS Coencapsulation of NO with Ar enabled us to adjust the amount of encapsulated NO. A total of 10 microl of gas can be encapsulated into 1 mg of liposomes. The release profile of NO from NO-ELIP demonstrated an initial rapid release followed by a slower release during the course of 8 h. Sixty-eight percent of cells remained viable when incubated with 80 microg/ml of NO/Ar-ELIP for 4 h. The delivery agent of NO to VSMCs by the use of NO/Ar-ELIP was 7-fold greater than unencapsulated NO. We discovered that NO/Ar-ELIP remained an effective delivery agent of NO to VSMCs even in the presence of hemoglobin. Local NO-ELIP administration to balloon-injured carotid arteries attenuated the development of intimal hyperplasia and reduced arterial wall thickening by 41 +/- 9%. CONCLUSIONS Liposomes can protect and deliver a bioactive gas to target tissues with the potential for both visualization of gas delivery and controlled therapeutic gas release.


Circulation | 2002

Left Ventricular Thrombus Enhancement After Intravenous Injection of Echogenic Immunoliposomes Studies in a New Experimental Model

Andrew J. Hamilton; Shaoling Huang; Drew Warnick; Adam C. Stein; Mark Rabbat; Taruna Madhav; Bonnie J. Kane; Ashwin Nagaraj; Melvin E. Klegerman; Robert C. MacDonald; David D. McPherson

Background—Targeted echogenic immunoliposomes (ELIPs) for ultrasound enhancement of atheroma components have been developed. To date, ELIP delivery has been intra-arterial. To determine whether ELIPs can be given intravenously with enhancement of systemic structures, a left ventricular thrombus (LVT) model was developed. Methods and Results—In 6 animals plus 1 dose-ranging animal, the apical coronary arteries were ligated, and an LVT was produced by injecting Hemaseel fibrin adhesive through the apical myocardium. The thrombus was imaged epicardially and transthoracically at 0, 1, 5, and 10 minutes after anti-fibrinogen ELIP injections. The dose of ELIPs was varied. PBS and unconjugated ELIPs were controls. The apical thrombi were easily reproduced and clearly visible with epicardial and transthoracic ultrasound. Enhancement occurred with 2 mg anti-fibrinogen ELIPs and increased with dose. With 8 mg ELIPs, enhancement was different from control within 10 minutes (P <0.05). Rhodamine-labeled anti-fibrinogen ELIPs were seen with fluorescence microscopy of the LVT. Blinded viewing detected enhancement by 10 minutes in all animals after anti-fibrinogen ELIPs. Conclusions—We describe an easily reproducible LVT model. Anti-fibrinogen ELIPs delivered intravenously, as a single-step process, rapidly enhance the ultrasound image of a systemic target. This allows for future development of ELIPs as a targeted ultrasound contrast agent.


Ultrasound in Medicine and Biology | 2002

Physical correlates of the ultrasonic reflectivity of lipid dispersions suitable as diagnostic contrast agents

Shaoling Huang; Andrew J. Hamilton; Edwin V. Pozharski; Ashwin Nagaraj; Melvin E. Klegerman; David D. McPherson; Robert C. MacDonald

The objective of this study was to determine the physical basis of ultrasound (US) reflectivity of echogenic lipid dispersions. These dispersions were made using a process previously described involving sonication of the lipid in water, addition of mannitol, freezing, lyophilization and rehydration. The component lipids were egg phosphatidylcholine, dipalmitoylphosphatidylethanolamine, dipalmitoylphosphatidylglycerol and cholesterol in a molar ratio of 69:8:8:15. Ultrasound reflectivity, as assessed with a 20-MHz intravascular US catheter and analyzed using computer-assisted videodensitometry, was found to be sensitive to variations in ambient pressure; echogenicity was greatly reduced by exposure to 0.5 atm vacuum for 10 s or 1.5 atm pressure for 10 s. Pressure changes of the magnitude that obtain in the arterial circulation had little effect on echogenicity. Vacuum treatment resulted in the release of approximately 100 microL air from a standard preparation of 10 mg lipid in 1 mL. Maximum ultrasonic reflectivity required the presence of 0.1-0.2 mol/L mannitol during the lyophilization step; mere addition of mannitol to the lipid lyophilized in the absence of mannitol produced nonreflective dispersions. Inclusion of sodium phosphate or other electrolytes reduced echogenicity. High echogenicity was associated with the presence of large-volume freeze-dried cakes and fusion of liposomes (which led to a 10 times increase in liposome diameters) during freezing before lyophilization. Lyophilization from water led to liposome fusion, but the cakes were small and US reflectivity was weak. Lyophilization from solutions of cryoprotectants such as trehalose produced large cakes, but little liposome fusion and also led to weak US reflectivity. Filtration through defined pores revealed that approximately 50% of the echogenicity originated from particles smaller than 1 microm and about 2/3 from particles smaller than 3 microm. These results indicate that lyophilization from 0.2 mol/L mannitol solution generates a disrupted array of lipid bilayers that, upon rehydration, fuse and trap small amounts of air distributed among liposome-size particles.


Ultrasound in Medicine and Biology | 2010

Ultrasound-Triggered Release of Recombinant Tissue-Type Plasminogen Activator from Echogenic Liposomes

Denise A. B. Smith; Sampada S. Vaidya; Jonathan A. Kopechek; Shaoling Huang; Melvin E. Klegerman; David D. McPherson; Christy K. Holland

Echogenic liposomes (ELIP) were developed as ultrasound-triggered targeted drug or gene delivery vehicles (Lanza et al. 1997; Huang et al. 2001). Recombinant tissue-type plasminogen activator (rt-PA), a thrombolytic, has been loaded into ELIP (Tiukinhoy-Laing et al. 2007). These vesicles have the potential to be used for ultrasound-enhanced thrombolysis in the treatment of acute ischemic stroke, myocardial infarction, deep vein thrombosis or pulmonary embolus. A clinical diagnostic ultrasound scanner (Philips HDI 5000; Philips Medical Systems, Bothell, WA, USA) equipped with a linear array transducer (L12-5) was employed for in vitro studies using rt-PA-loaded ELIP (T-ELIP). The goal of this study was to quantify ultrasound-triggered drug release from rt-PA-loaded echogenic liposomes. T-ELIP samples were exposed to 6.9-MHz B-mode pulses at a low pressure amplitude (600 kPa) to track the echogenicity over time under four experimental conditions: (1) flow alone to monitor gas diffusion from the T-ELIP, (2) pulsed 6.0-MHz color Doppler exposure above the acoustically driven threshold (0.8 MPa) to force gas out of the liposome gently, (3) pulsed 6.0-MHz color Doppler above the rapid fragmentation threshold (2.6 MPa) or (4) Triton X-100 to rupture the T-ELIP chemically as a positive control. Release of rt-PA for each ultrasound exposure protocol was assayed spectrophotometrically. T-ELIP were echogenic in the flow model (5 mL/min) for 30 min. The thrombolytic drug remained associated with the liposome when exposed to low-amplitude B-mode pulses over 60 min and was released when exposed to color Doppler pulses or Triton X-100. The rt-PA released from the liposomes had similar enzymatic activity as the free drug. These T-ELIP are robust and echogenic during continuous fundamental 6.9-MHz B-mode imaging at a low exposure output level (600 kPa). Furthermore, a therapeutic concentration of rt-PA can be released by fragmenting the T-ELIP with pulsed 6.0-MHz color Doppler ultrasound above the rapid fragmentation threshold (1.59 MPa). (E-mail: [email protected]).


Journal of the Acoustical Society of America | 2012

Passive imaging with pulsed ultrasound insonations

Kevin J. Haworth; T. Douglas Mast; Kirthi Radhakrishnan; Mark T. Burgess; Jonathan A. Kopechek; Shaoling Huang; David D. McPherson; Christy K. Holland

Previously, passive cavitation imaging has been described in the context of continuous-wave high-intensity focused ultrasound thermal ablation. However, the technique has potential use as a feedback mechanism for pulsed-wave therapies, such as ultrasound-mediated drug delivery. In this paper, results of experiments and simulations are reported to demonstrate the feasibility of passive cavitation imaging using pulsed ultrasound insonations and how the images depend on pulsed ultrasound parameters. The passive cavitation images were formed from channel data that was beamformed in the frequency domain. Experiments were performed in an invitro flow phantom with an experimental echo contrast agent, echogenic liposomes, as cavitation nuclei. It was found that the pulse duration and envelope have minimal impact on the image resolution achieved. The passive cavitation image amplitude scales linearly with the cavitation emission energy. Cavitation images for both stable and inertial cavitation can be obtained from the same received data set.


Journal of Ultrasound in Medicine | 2008

Ultrasound-Mediated Release of Hydrophilic and Lipophilic Agents From Echogenic Liposomes

Jonathan A. Kopechek; Todd M. Abruzzo; Boyu Wang; Stephen M. Chrzanowski; Denise A. B. Smith; Patrick Kee; Shaoling Huang; Joel H. Collier; David D. McPherson; Christy K. Holland

Objective. To achieve ultrasound‐controlled drug delivery using echogenic liposomes (ELIPs), we assessed ultrasound‐triggered release of hydrophilic and lipophilic agents in vitro using color Doppler ultrasound delivered with a clinical 6‐MHz compact linear array transducer. Methods. Calcein, a hydrophilic agent, and papaverine, a lipophilic agent, were each separately loaded into ELIPs. Calcein‐loaded ELIP (C‐ELIP) and papaverine‐loaded ELIP (P‐ELIP) solutions were circulated in a flow model and treated with 6‐MHz color Doppler ultrasound or Triton X‐100. Treatment with Triton X‐100 was used to release the encapsulated calcein or papaverine content completely. The free calcein concentration in the solution was measured directly by spectrofluorimetry. The free papaverine in the solution was separated from liposome‐bound papaverine by spin column filtration, and the resulting papaverine concentration was measured directly by absorbance spectrophotometry. Dynamic changes in echogenicity were assessed with low‐output B‐mode ultrasound (mechanical index, 0.04) as mean digital intensity. Results. Color Doppler ultrasound caused calcein release from C‐ELIPs compared with flow alone (P < .05) but did not induce papaverine release from P‐ELIPs compared with flow alone (P > .05). Triton X‐100 completely released liposome‐associated calcein and papaverine. Initial echogenicity was higher for C‐ELIPs than P‐ELIPs. Color Doppler ultrasound and Triton X‐100 treatments reduced echogenicity for both C‐ELIPs and P‐ELIPs (P < .05). Conclusions. The differential efficiency of ultrasound‐mediated pharmaceutical release from ELIPs for water‐ and lipid‐soluble compounds suggests that water‐soluble drugs are better candidates for the design and development of ELIP‐based ultrasound‐controlled drug delivery systems.


Ultrasound in Medicine and Biology | 2008

A method to co-encapsulate gas and drugs in liposomes for ultrasound-controlled drug delivery.

Shaoling Huang; David D. McPherson; Robert C. MacDonald

We describe a novel method for the facile production of gas-containing liposomes with simultaneous drug encapsulation. Liposomes of phospholipid and cholesterol were prepared by conventional procedures of hydrating the lipid film, sonicating, freezing and thawing. A single but critical modification of this procedure generates liposomes that contain gas (air, perfluorocarbon, argon); after sonication, the lipid is placed under pressure with the gas of interest. After equilibration, the sample is frozen. The pressure is then reduced to atmospheric and the suspension thawed. This procedure leads to entrapment of air in amounts up to 10% by volume in lipid dispersions at moderate (10 mg/mL) concentrations of lipids. The amount of gas encapsulated increases with gas pressure and lipid concentration. Using 0.32 mol/L mannitol to provide an aqueous phase with physiological osmolarity, 1, 3, 6 or 9 atm of pressure was applied to 4 mg of lipid. This led to encapsulation of 10, 15, 20 and 30 microl of gas in a total of 400 microl of liposome dispersion (10 mg lipids/mL), respectively. The mechanism for gas encapsulation presumably depends on the fact that air (predominantly nitrogen and oxygen), like most solutes, dissolves poorly in ice and is excluded from the ice that forms during freezing. The excluded air then comes out of solution as air pockets that are stabilized in some form by a lipid coating. The presence of air in these preparations sensitizes them to ultrasound (1MHz, 8 W/cm2,10 s) such that up to half of their aqueous contents (which could be a water soluble drug) can be released by short (10 s) applications of ultrasound. Both diagnostic and therapeutic applications of the method are conceivable.


Journal of Drug Targeting | 2007

Fibrin targeting of tissue plasminogen activator-loaded echogenic liposomes

Susan D. Tiukinhoy-Laing; Kyle D. Buchanan; Devang Parikh; Shaoling Huang; Robert C. MacDonald; David D. McPherson; Melvin E. Klegerman

We recently reported entrapment of tissue-plasminogen activator (tPA) into echogenic liposomes (ELIP) with retention of echogenicity and thrombolytic effect. Integral to the potential of this agent for ultrasound-detectable local drug delivery is the specific binding of tPA–ELIP to clots. tPA contains fibrin-binding sites; we hypothesized that tPA when associated with ELIP, will maintain fibrin binding properties, rendering further manipulation for targeting of the tPA–ELIP unnecessary. We demonstrated strong fibrin binding of the ELIP-associated tPA. Fibrin binding for ELIP-associated tPA was twice that of free tPA. This strong affinity for fibrin was confirmed using echogenicity analysis of porcine clots in vitro. Both objective (mean gray scale analysis) and subjective (visual estimation by two experienced echocardiographers) evaluation of the clots showed enhanced highlighting of clots treated with tPA–ELIP when compared to control. The findings in this study represent new approaches for fibrin-targeted, ultrasound-directed and enhanced local delivery of a thrombolytic agent.

Collaboration


Dive into the Shaoling Huang's collaboration.

Top Co-Authors

Avatar

David D. McPherson

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melvin E. Klegerman

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Hyunggun Kim

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tao Peng

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Melanie R. Moody

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Kee

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge